Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf ·...

28
Leber Hereditary Optic Neuropathy: A Mitochondrial Disease Unique in Many Ways Rui Bi, Ian Logan, and Yong-Gang Yao Contents 1 LHON Clinical Features .................................................................... 311 2 Epidemiology ................................................................................ 312 3 mtDNA Primary Mutations ................................................................. 312 3.1 Three Well-Known Primary Mutations .............................................. 312 3.2 Rare Primary Mutations ............................................................... 314 3.3 Mutation Heteroplasmy and Occurrence of Double Primary LHON Mutations .... 315 4 Effect of Secondary Mutations and mtDNA Genetic Background ........................ 316 4.1 The Secondary Mutations May Act in Synergy with the Primary Mutations ....... 316 4.2 mtDNA Haplogroup Background Effect ............................................. 317 4.3 Occurrence of mtDNA Pathogenic Mutations with the Primary Mutations ........ 319 4.4 mtDNA Mutational Hotspots in Suspected LHON Patients ......................... 319 5 Effect of Nuclear Genes on LHON ......................................................... 320 5.1 Chromosome X ........................................................................ 320 5.2 Other Nuclear Factors ................................................................. 320 6 Environmental Factors ...................................................................... 321 7 Biochemical Effects of mtDNA Mutations and Their Implication for LHON Treatment 322 7.1 Biochemical Effect of mtDNA Mutations ............................................ 322 7.2 Functional Characterization of the LHON Primary Mutations ...................... 323 7.3 New Avenues of Research: LHON Gene Therapy and Genome Editing ........... 324 7.4 Implications for Medical Therapy for LHON ........................................ 325 8 Conclusion and Future Perspective ......................................................... 326 References ....................................................................................... 326 R. Bi • Y.-G. Yao (*) Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China e-mail: [email protected] I. Logan Exmouth, Devon, UK # Springer International Publishing Switzerland 2016 H. Singh, S.-S. Sheu (eds.), Pharmacology of Mitochondria, Handbook of Experimental Pharmacology 240, DOI 10.1007/164_2016_1 309

Transcript of Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf ·...

Page 1: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

Leber Hereditary Optic Neuropathy:A Mitochondrial Disease Uniquein Many Ways

Rui Bi, Ian Logan, and Yong-Gang Yao

Contents

1 LHON Clinical Features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 311

2 Epidemiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 312

3 mtDNA Primary Mutations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 312

3.1 Three Well-Known Primary Mutations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 312

3.2 Rare Primary Mutations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 314

3.3 Mutation Heteroplasmy and Occurrence of Double Primary LHON Mutations . . . . 315

4 Effect of Secondary Mutations and mtDNA Genetic Background . . . . . . . . . . . . . . . . . . . . . . . . 316

4.1 The Secondary Mutations May Act in Synergy with the Primary Mutations . . . . . . . 316

4.2 mtDNA Haplogroup Background Effect . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 317

4.3 Occurrence of mtDNA Pathogenic Mutations with the Primary Mutations . . . . . . . . 319

4.4 mtDNA Mutational Hotspots in Suspected LHON Patients . . . . . . . . . . . . . . . . . . . . . . . . . 319

5 Effect of Nuclear Genes on LHON . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 320

5.1 Chromosome X . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 320

5.2 Other Nuclear Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 320

6 Environmental Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 321

7 Biochemical Effects of mtDNA Mutations and Their Implication for LHON Treatment 322

7.1 Biochemical Effect of mtDNA Mutations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 322

7.2 Functional Characterization of the LHON Primary Mutations . . . . . . . . . . . . . . . . . . . . . . 323

7.3 New Avenues of Research: LHON Gene Therapy and Genome Editing . . . . . . . . . . . 324

7.4 Implications for Medical Therapy for LHON . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 325

8 Conclusion and Future Perspective . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 326

References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 326

R. Bi • Y.-G. Yao (*)

Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal Models and

Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences,

Kunming, Yunnan, China

e-mail: [email protected]

I. Logan

Exmouth, Devon, UK

# Springer International Publishing Switzerland 2016

H. Singh, S.-S. Sheu (eds.), Pharmacology of Mitochondria,Handbook of Experimental Pharmacology 240, DOI 10.1007/164_2016_1

309

Page 2: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

Abstract

Leber hereditary optic neuropathy (LHON) was the first mitochondrial disease to

be identified as being caused by mutations in the mitochondrial DNA (mtDNA).

This disease has been studied extensively in the past two decades, particularly in

Brazilian, Chinese and European populations; and many primary mutations have

been reported. However, the disease is enigmatic with many unique features, and

there still are several important questions to be resolved. The incomplete pene-

trance, the male-biased disease expression and the prevalence in young adults all

defy a proper explanation. It has been reported that the development of LHON is

affected by the interaction between mtDNA mutations, mtDNA haplogroup

background, nuclear genes, environmental factors and epigenetics. Furthermore,

with the help of new animal models for LHON that have been created in recent

years, we are continuing to learn more about the mechanism of this disease. The

stage has now been reached at which there is a good understanding of both the

genetic basis of the disease and its epidemiology, but just how the blindness that

follows from the death of cells in the optic nerve can be prevented remains to be

a pharmacological challenge. In this chapter, we summarize the progress that has

been made in various recent studies on LHON, focusing on the molecular

pathogenic mechanisms, clinical features, biochemical effects, the pharmacol-

ogy and its treatment.

Keywords

Animal model • LHON • mtDNA • Nuclear genes • Therapy

Leber hereditary optic neuropathy (LHON, MIM 535000) is a common cause of

acute or subacute central visual loss in young adults, with a high male-to-female

ratio for the clinical expression of symptoms (Carelli et al. 2004; Man et al. 2002b;

Yen et al. 2006; Yu-Wai-Man et al. 2009). LHON was first described by German

ophthalmologist Theodore Leber in 1871 (Erickson 1972; Leber 1871) and was

later recognized to be a mitochondrial disease (Wallace et al. 1988). In the past two

decades, our knowledge about this disease was mainly derived from studies involv-

ing patients with European roots; although it is generally considered the disease

occurs worldwide. We now know that the majority of LHON cases are caused by

the presence of one of three primary mitochondrial DNA (mtDNA) mutations

(m.11778G>A, m.14484T>C and m.3460G>A) (Howell et al. 1991; Huoponen

et al. 1991; Mackey and Howell 1992; Wallace et al. 1988). From the 1990s,

researchers all around the world have carried out large number of studies to

characterize the genetic and clinical features of LHON (Carelli et al. 2004; Ji

et al. 2008; Man et al. 2002b; Yen et al. 2006; Yu-Wai-Man et al. 2009; Yu

et al. 2010b; Zhang et al. 2009). These studies have greatly increased our under-

standing of the pathology and risk factors of LHON. However, the two major

features of the disease, incomplete penetrance (not all mutation carriers demon-

strate clinical expression of disease) and gender bias, still are the most urgent

310 R. Bi et al.

Page 3: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

challenges for understanding the pathogenesis of this disease (Carelli et al. 2004;

Man et al. 2002b; Yen et al. 2006; Yu-Wai-Man et al. 2009). Furthermore, medical

treatment for this disease has still not been satisfactorily resolved (La Morgia

et al. 2014).

1 LHON Clinical Features

The most typical characteristics of patients with LHON are the following: (1) a

family history that shows a matrilineal inheritance pattern, although there are a

considerable number of sporadic LHON cases; (2) the visual loss is painless,

affecting just one eye initially and then involving the other eye and going on to

cause a significant loss of vision or even total blindness, within a short time; and

(3) more male carriers of the pathogenic mutations develop symptoms of LHON

than maternally related female carriers of the same primary LHON mutations.

About 50% of the male carriers and 10% of the female carriers will be affected,

while males from 15 to 35 years old are the main affected population (Seedorff

1985). The progress of LHON can be divided into an acute phase and a chronic

phase based on the clinical features. In the majority of patients, the deterioration of

vision is acute and affects the two eyes simultaneously (25%) or sequentially (75%)

within about 8 weeks (Harding et al. 1995). This stage of the disease is associated

with retinal vascular engorgement, deformation of the optic nerve and oedema of

the retina (Yen et al. 2006), while there also are some patients with no pathological

changes to be observed (Riordan-Eva and Harding 1995). In addition, the disease

progression may present slowly in some patients, lasting for 6 months or more. In

this kind of patient, the visual field changes are not significant, thus increasing the

difficulty of diagnosis (Nikoskelainen et al. 1996), particularly in the cases which

lack the typical family history of LHON. In addition to typical LHON symptoms,

the patient may also suffer from other problems, such as muscle atrophy, multiple

sclerosis, heart disease and cerebrovascular abnormalities (Jaros et al. 2007; La

Morgia et al. 2008; Sorajja et al. 2003).

Because the clinical manifestations of LHON vary widely, patients with compli-

cated symptoms are often not diagnosed with LHON accurately. An important

diagnostic marker for LHON is the matrilineal inheritance pattern, but for those

patients with the clinical features of LHON but who lack a family history, screening

the mtDNA for the three primary mutations (m.3460G>A, m.11778G>A and

m.14484T>C) is necessary for an accurate diagnosis (Jia et al. 2006). It is worth

noting that there are many suspected LHON patients who have symptoms but no

family history and with no known primary mutations, and this suggests other

genetic or environmental factors may influence the onset of LHON, or produce

similar, and as yet unidentified, diseases (Carelli et al. 2004; Kirkman et al. 2009;

Yen et al. 2006).

Leber Hereditary Optic Neuropathy: A Mitochondrial Disease Unique in Many Ways 311

Page 4: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

2 Epidemiology

The reported prevalence of LHON varied among European countries: about

1/25,000 in the north-east of England (Chinnery et al. 2000; Man et al. 2003),

1/39,000 in the Netherlands (Spruijt et al. 2006) and 1/50,000 in Finland (Puomila

et al. 2007). But in China and other regions of Asia, there has not been a compre-

hensive epidemiological survey. In 2008, Elliott et al. screened for LHON primary

mutations in the neonatal umbilical cord blood samples of 3,000 normal newborns

and identified a relatively high frequency of 0.107% for m.3460G>A (3/2807),

0.108% for m.11778G>A (3/2770) and 0.105% for m.14484T>C (3/2855) (Elliott

et al. 2008). This study indicated that the primary LHON mutations do exist in

normal populations. We screened for the three LHON primary mutations by using a

sensitive allele-specific PCR technique in the general Han Chinese population

(1,555 healthy samples), but found no LHON primary mutation carriers, suggesting

that the frequency of the LHON primary mutation in Han Chinese general

populations is very low (Bi et al. 2010). Large-scale epidemiological surveys for

LHON still need to be carried out in Asian populations and populations from other

continents to get a complete profile of LHON prevalence.

3 mtDNA Primary Mutations

3.1 Three Well-Known Primary Mutations

In 1988, Wallace and his co-workers were the first to propose an mtDNA mutation

as the cause of LHON (Wallace et al. 1988). Since then many mtDNA mutations

have been reported as being LHON primary mutations (cf. http://www.mitomap.

org) (Fig. 1). Among these mutations, three mutations (m.3460G>A in the

MT-ND1 gene, m.11778G>A in the MT-ND4 gene and m.14484T>C in the

MT-ND6 gene) are reported to account for more than 95% of the LHON patients

(Mackey et al. 1996).

These three primary mutations show considerable variation in their levels of

evolutionary conservation. Mutation m.11778A>G is highly conserved, while

mutations m.3460G>A and m.14484T>C are less conserved, which suggests that

not all pathogenic mutations occur in highly conserved regions. The frequency of

these three mutations varies among different geographical regions. In Europe,

mutation m.11778G>A is found in 56.6% of patients, and mutations

m.3460G>A and m.14484T>C are found in 22.6 and 20.8% of patients, respec-

tively (Hudson et al. 2007b). In Chinese populations, the frequency of

m.11778G>A is much higher than that of European populations and populations

of European origin and can be up to 90.2%. The frequencies of m.3460G>A and

m.14484T>C are much lower than those of European populations and populations

of European origin, with a frequency of 1.1 and 8.7%, respectively (Ji et al. 2008;

Jia et al. 2006; Yu et al. 2010a,b) (Fig. 2). A founder effect (the fixation of an allele

after the migration and population expansion of a small population in a new area)

312 R. Bi et al.

Page 5: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

D‐loop

TF 12S rRNA

TV

16S rRNA

TL1

ND1

TITQTM

ND2TW

TATNTCTY

CO1TS1TD

CO2

TKATP8/ATP6

CO3

TGND3

TRND4L

ND4

THTS2TL2

ND5

ND6TE

CYB

TT TP

control regiontRNArRNAcomplex Icomplex IIIcomplex IVATP synthase

m.3460G>Ap.A52T

m.14484T>Cp.M64V

m.3635G>Ap.S110N

m.14459G>Ap.A72V

m.3700G>Ap.A112T

m.3733G>Ap.E143K

m.14568T>Cp.G36S

m.10680G>Ap.A71T

m.11778G>Ap.R340H

m.4171C>Ap.L289M

Fig. 1 Schematic profile of LHON mutations. The three primary mutations and the other rare

mtDNA mutations that have been reported in at least three independent LHON families, as

summarized in MITOMAP, http://www.mitomap.org/bin/view.pl/MITOMAP/MutationsLHON,

are shown in their relative positions on the mtDNA molecule (Achilli et al. 2012; Brown

et al. 2001b; Bu and Rotter 1991; Fauser et al. 2002a; Fauser et al. 2002b; Gropman et al. 2004;

Horvath et al. 2002; Kim et al. 2002; Shoffner et al. 1995; Valentino et al. 2004; Wissinger

et al. 1997; Zhang et al. 2012)

m.3460G>A22.6%

m.14484T>C20.8%

m.11778G>A56.6%

m.14484T>C8.7%

m.3460G>A1.1%

m.11778G>A90.2%

European population Chinese population N = 159 families N = 537 families

Fig. 2 Mutation spectra of known primary LHON mutations in Chinese and European

populations. Frequency of the three LHON primary mutations in European (left) (Hudson

et al. 2007b) and Han Chinese (right) populations (Ji et al. 2008; Jia et al. 2006; Yu

et al. 2010a, b)

Leber Hereditary Optic Neuropathy: A Mitochondrial Disease Unique in Many Ways 313

Page 6: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

was observed for m.14484T>C in Canada, which accounts for 87% of the cases and

appears to have had an origin in France (Macmillan et al. 2000; Macmillan

et al. 1998). However, the best founder effect has been reported from Brazil,

where many hundreds of persons can trace their maternal ancestry and the carriage

of mutation m.11778G>A, back to an immigrant family that came from Italy

(Sadun et al. 2002). The different frequencies of the three primary mutations in

different populations may be caused by the past demographic history.

3.2 Rare Primary Mutations

In addition to the three primary mutations, some rare mutations, i.e. m.3635G>A,

m.3700G>A, m.14459G>A and so on, as summarized in MITOMAP (http://www.

mitomap.org/bin/view.pl/MITOMAP/MutationsLHON) from previous studies

(Achilli et al. 2012; Brown et al. 2001b; Fauser et al. 2002b; Gropman et al. 2004;

Horvath et al. 2002; Shoffner et al. 1995) have also been reported (Fig. 1). Many of

these rare primary mutations were reported in single patients with the typical

features of LHON. We have shown how we identified mutations m.3635G>A

and m.10680G>A as being pathogenic as follows.

Mutation m.3635G>A was first identified in a Russian LHON family (Brown

et al. 2001b) and was confirmed to cause dysfunction of the mitochondrial

respiratory chain. Recently, we identified mutation m.3635G>A in a family that

had no known primary mutation but had a clear matrilineal inheritance and typical

clinical pattern of LHON (Zhang et al. 2009). At about the same time, Yang

et al. (2009a) also identified m.3635G>A in two Chinese LHON families. Subse-

quently, Jia et al. (2010) screened for this mutation in a large sample set of

suspected LHON patients who showed the clinical features of LHON but without

any of the three primary mutations and identified eight families with the

m.3635G>A mutation. These authors further characterized the clinical symptoms

of the representative probands with the mutation and provided evidence for a

causal association of m.3635G>A with LHON (Jia et al. 2010). We analyzed the

complete mtDNA genome sequences of these families and found that

m.3635G>A had multiple origins in Chinese (Bi et al. 2012). Furthermore,

m.3635G>A is highly conserved in vertebrates, suggesting the importance of

this site in the MT-ND1 protein (Zhang et al. 2009). Recently, mutation

m.3635G>A was again reported in nine Chinese families and one family from

Poland (Kodron et al. 2014; Zhang et al. 2014). Therefore, we can now propose

that the mutation m.3635G>A does cause LHON and has a frequency similar to

the m.3460G>A mutation (0.57 vs. 0.50%) in Chinese LHON patients

(Bi et al. 2012; Jia et al. 2010; Zhang et al. 2009). This mutation should be

considered routinely when testing Chinese patients with the symptoms and signs

of LHON disease.

Mutation m.10680G>A in the MT-ND4L gene can be given as another good

example for the identification of additional LHON mutations. This mutation was

314 R. Bi et al.

Page 7: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

first reported in a Chinese family in which all the members had a partial reduction in

their vision or had complete blindness, while the mutation was not found in

unaffected family members, nor in 100 controls (Yang et al. 2009b). Through

complete mitochondrial genome sequencing, they found that the matrilineal family

members had the primary mutation m.14484T>C and one novel mutation

m.10680G>A. Accordingly, Yang et al. (2009b) suggested that mutation

m.10680G>A might be the reason for the complete penetrance of LHON

symptoms in this family, although this symptom severity may also be caused by

m.14484T>C. Later on, we identified mutation m.10680G>A in one suspected

LHON family without any of the three well-known primary mutations and

performed an analysis from an evolutionary perspective which suggested a poten-

tially pathogenic role of m.10680G>A in LHON (Zou et al. 2010). Recently, we

optimized an allele-specific PCR technique to screen for m.10680G>A in

774 patients with suspected LHON and identified two patients with the mutation

(Zhang et al. 2012). The predicted protein structure revealed that mutation

m.10680G>A may lead to LHON by changing the structure of the transmembrane

region in the MT-ND4L protein (Zhang et al. 2012). However, there remains to be a

doubt concerning whether this mutation is significant outside China, as there are

eight mitochondrial sequences in the GenBank database from other parts of the

world which show this mutation but are not specifically linked to LHON disease

(data searched on June 25, 2015).

3.3 Mutation Heteroplasmy and Occurrence of Double PrimaryLHON Mutations

Depending on its particular type, a cell may contain hundreds or thousands of

mitochondria. The presence of a mutation only in some of cells of an individual or

in some of the mitochondria within a cell indicates heteroplasmy for that mutation.

The existence of wild-type and mutant mtDNAwithin cells (intracell heteroplasmy)

and among cells (intercell heteroplasmy) leads to different levels of mutation load

(0–100%) within a cell or tissue (Yao et al. 2015). The heteroplasmy level of a

pathogenic mutation may affect the onset of a mitochondrial disease, and there is a

threshold effect (DiMauro and Schon 2003). Most families with LHON primary

mutation are homoplasmic, but about 10–15% of families do show heteroplasmy

(Carelli et al. 2004; Man et al. 2002b; Yen et al. 2006; Yu-Wai-Man et al. 2009).

However, a high prevalence of LHON family with heteroplasmic m.11778G>A

(11 in 30 pedigrees, 37%) was observed in Thai population (Phasukkijwatana

et al. 2006). The level of heteroplasmy affects the presence of LHON symptoms,

and it is reported that when the level of heteroplasmy is less than 60%, there is a

lower probability of disease (Chinnery et al. 2001a). But there are also some reports

showing cases with disease symptoms even if the heteroplasmy is at a low level,

suggesting a modulation effect from the nuclear genetic background (Black

et al. 1996; Jacobi et al. 2001).

Leber Hereditary Optic Neuropathy: A Mitochondrial Disease Unique in Many Ways 315

Page 8: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

In our previous studies, we have found that only a small part of Chinese LHON

patients (about 0.2% of patients with m.11778G>A and 5.8% of patients with

m.14484T>C) carried heteroplasmic primary mutations, while most of the patients

were homoplasmic (Yu et al. 2010b; Zhang et al. 2011b). The pattern of extremely

low frequency of heteroplasmic m.11778G>A in Chinese patients appears to be

different from that of European patients or patients of European origin. In six

LHON families with the mutation m.3460G>A that we analyzed, we only

identified one family with a heteroplasmic mutation, and the proband of this family

had a heteroplasmic mutation load of about 40%. Interestingly, the penetrance of

LHON symptoms in this family was 12.5%, which was significantly lower than the

average penetrance of families (25.6%) with a homoplasmic mutation

(Yu et al. 2010a). The heteroplasmy status of m.3460G>A may be one of the

reasons for the low penetrance of LHON in this family (Yu et al. 2010a), which is

consistent with previous findings that only 20% of heteroplasmic carriers, but 47%

of homoplasmic carriers, manifested the disease in a Thai population

(Phasukkijwatana et al. 2006).

In addition, there are sporadic reports of the presence of two LHONmutations in

one family. Brown et al. (2001a) reported a 9-year-old Caucasian girl carrying both

the m.11778G>A and m.14484T>C mutations, with m.11778G>A in a

heteroplasmic status. Despite carrying two pathogenic mutations, the patient’s

mother was apparently normal (Brown et al. 2001a). Tonska et al. (2008) reported

a Polish LHON family with both the m.11778G>A and m.3460G>A mutations,

with m.3460G>A in a heteroplasmic status. Except for typical LHON symptoms,

no other nervous system disorders were found in this family, and all the female

members were unaffected (Tonska et al. 2008). These studies seem to indicate that

heteroplasmic mutations may be linked to fewer manifestations of LHON.

4 Effect of Secondary Mutations and mtDNA GeneticBackground

According to the clinical observation, different families present a variable pene-

trance of LHON symptoms, ranging from 10 to 100%, albeit harbouring the same

primary mutation, as shown by the two families with m.11778G>A in Han Chinese

(Wang et al. 2008). Secondary mtDNA mutations and mtDNA haplogroup genetic

background appear to have some effect on the penetrance of LHON. These two

factors may not lead to LHON directly, but can act in synergy with primary

mutations to affect the onset of LHON.

4.1 The Secondary Mutations May Act in Synergywith the Primary Mutations

Most of the secondary mutations are common variants. For instance, variant

m.12811T>C, which is the defining variants of haplogroup M7b102 and has a

316 R. Bi et al.

Page 9: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

high frequency in the general population (Kong et al. 2006), was considered to be

one of the secondary mutations of LHON (Huoponen et al. 1993). We have

obtained consistent results to show that haplogroup M7b102 is an important risk

factor for the development of LHON in Han Chinese patients with m.11778G>A

mutation (Ji et al. 2008; Zhang et al. 2011b). Intriguingly, m.12811T>C also

defined a clade of H3h in European populations or populations of European origin,

which also shows an increased prevalence of LHON (Bandelt H-J et al. unpublished

data). All these results suggested m.12811T>C as being a secondary mutation for

LHON, both in Chinese and European populations. Also the mutation m.593T>C is

a common variant found in the MT-tRNAPhe gene. Our recent study revealed that

this variant may lead to altered structure of MT-tRNAPhe and thus acts in synergy

with m.11778G>A to increase the penetrance of LHON (Zhang et al. 2011a). Note

that many of the secondary mutations of LHON are haplogroup-defining variants

and have a much higher frequency than the primary LHON mutations. Just why

some of these secondary mutations are associated with LHON only in some

populations, but not others, is unresolved.

4.2 mtDNA Haplogroup Background Effect

Because of high mutation rate and lack of recombination, mtDNA accumulates

mutations in a time order within a relatively short span of time and forms different

haplotypes (Pakendorf and Stoneking 2005). In the process of human evolution and

migration, descendants of an original mtDNA haplotype would sequentially accu-

mulate additional mutations to generate a group of related haplotypes that was

termed as a haplogroup (Achilli et al. 2008; Kong et al. 2006; Torroni et al. 2000).

The current global mtDNA phylogenetic tree contains over 4,000 different

haplogroups, which form a region-specific distribution pattern (Achilli

et al. 2008; Kong et al. 2010; Kong et al. 2006; van Oven and Kayser 2009).

mtDNA haplogroups have become important genetic markers for different

populations. In the past decades, mtDNA haplogroups have also been identified

to be associated with many different metabolic and neurodegenerative diseases. As

early as in 1992, Brown et al. (1992) found that mtDNA haplotype (5244-13708-

15257-15812) was associated with significantly increased risk for the onset of

LHON. Later, Torroni et al. (1997) analyzed mtDNA haplogroup in Italian

LHON patients and identified haplogroup J as a risk factor for patients with

m.11778G>A and m.14484T>C. Shafa Shariat Panahi et al. (2006) found that

haplogroups J and W increased disease risk in Iranian LHON patients carrying

m.11778G>A and m.3460G>A, respectively. However, the sample size of these

studies was too small to obtain a firm conclusion. In recent years, further studies

have conducted to investigate the association between mtDNA haplogroup and

LHON. One of the most comprehensive studies was carried out by Hudson et al.

(2007b), who analyzed mtDNA haplogroups in 3,613 individuals from 159 -

European LHON families and found that haplogroups J2, J1 and K increased the

LHON risk in patients with m.11778G>A, m.14484T>C and m.3460G>A,

Leber Hereditary Optic Neuropathy: A Mitochondrial Disease Unique in Many Ways 317

Page 10: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

respectively, while haplogroup H was a protective factor for patients with

m.11778G>A. Phylogenetic analysis indicated that the nonsynonymous variants

located in the root of haplogroups J and K may account for the association between

these haplogroups and LHON (Hudson et al. 2007b).

Because the genetic structure is different between Asian and European

populations, we have performed a comprehensive study, with the largest number

of LHON patients in the Han Chinese population including 1,859 individuals from

182 Chinese families (including one from Cambodia) with the m.11778G>A

mutation, to investigate whether mtDNA haplogroups affect the penetrance of

LHON in East Asian families. Haplogroup M7b102 was identified to be signifi-

cantly associated with visual loss, whereas haplogroup M8a showed a protective

effect (Ji et al. 2008). Further analyses of the complete mtDNA sequences from

LHON families with m.11778G>A suggested that variants m.12811T>C

(p.Y159H) in the MT-ND5 gene and m.8584G>A – m.8684C>T (p.A20T – p.T53I)

in the MT-ATP6 gene, which are haplogroup-defining variants for haplogroups

M7b102 and M8a, respectively, might account for the association of these two

haplogroups with LHON (Ji et al. 2008). Subsequently, we analyzed the matrilineal

structure of further 304 Chinese patients with m.11778G>A and 843 patients with

suspected LHON, to determine the effect of the mtDNA genetic background on the

onset of disease (Zhang et al. 2011b). The enlarged sample size of LHON patients

confirmed the results that were obtained in our previous study based on 182 families

(Ji et al. 2008). Specifically, the LHON samples differed from the general Chinese

or samples with suspected LHON by harbouring fewer lineages belonging to

haplogroup F and more lineages belonging to M7b and D4. In contrast, the

matrilineal structure of the suspected LHON population resembled that of the

general Chinese population, suggesting no role of mtDNA haplogroup in these

suspected LHON patients (Zhang et al. 2011b). In Southeast Asian population,

haplogroup B5a1 was identified to be significantly associated with m.11778G>A

and appeared to modify the risk of visual loss (Kaewsutthi et al. 2011).

Although association studies have indicated that the onset of LHON is

influenced by mtDNA haplogroup, functional characterization of this effect is a

daunting task. There have been several attempts that have increased our knowledge

regarding the background effect. For instance, Suissa et al. (2009) found that

ancient mtDNA variants of haplogroup J reduced the replication and stability of

mtDNA. Using cybrid cells with different mtDNA haplogroup backgrounds, Ghelli

et al. (2009) found that haplogroup J increases the sensitivity of cells to

2,5-hexanedione toxicity, indicating that the mtDNA haplogroup background

affects the development of LHON in combination with environmental factors.

However, tests of respiratory function in cybrid cell lines carrying European

mtDNA haplogroups (including J) that were demonstrated to be associated with

LHON showed no detectable differences in multiple parameters, suggesting that the

effect of the mtDNA haplogroup may be more complex than what we had thought

(Carelli et al. 2002).

Note that a search of the 30,000 mtDNA sequences (data searched on June 25,

2015) held by the GenBank database shows that many of the European sequences

318 R. Bi et al.

Page 11: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

containing a primary LHON mutation do come from the subgroups of

haplogroup U, a feature which as yet has not raised the attention of researchers.

4.3 Occurrence of mtDNA Pathogenic Mutationswith the Primary Mutations

Besides common variants, there were some pathogenic mutations for other mito-

chondrial diseases coexisting with the LHON primary mutations. Mimaki et al.

(2003) reported one 51-year-old Japanese patient carrying both m.11778G>A and

m.12192G>A (a pathogenic mutation for cardiomyopathy). The initial symptom of

this patient was rapid reduction of weight, with acute visual loss developing

6 months later. We reported a LHON family with two pathogenic mutations

m.11778G>A and m.1555A>G, which may account for the high penetrance

(78.6%) of LHON in this family (Zhang et al. 2008). Mutation m.1555A>G is

the major cause of aminoglycoside-induced and non-syndromic hearing loss (Guan

2011; Kokotas et al. 2007; Petit et al. 2001). However, none of the matrilineal

relatives showed problems with their hearing, and we therefore inferred that the

mutation m.1555A>G in this family may act in synergy with m.11778G>A to

significantly increase the penetrance of LHON (Zhang et al. 2008). Interestingly,

the co-occurrence of m.1555A>G and m.11778G>A was also observed in two

Indian families recently, with one family showing a high penetrance of LHON

(62.5%) (Khan et al. 2013).

4.4 mtDNA Mutational Hotspots in Suspected LHON Patients

Though most of the LHON patients have been identified to carry known mtDNA

primary mutations, there still are many patients with typical clinical features of

LHON but without known mtDNA primary mutations. These patients have not

been fully investigated, and genetic analysis has revealed that there might be

mtDNA mutation hotspots in these patients. Studies in European populations

revealed that the MT-ND1 and MT-ND6 genes were mutational hotspots for

nonsynonymous mutations (Chinnery et al. 2001b; Valentino et al. 2004), whereas

in our study, albeit with a limited number of Han Chinese families, we showed that

theMT-ND1 andMT-ND5 genes are mutational hotspots in families with suspected

LHON (Zou et al. 2010). However, it remains to be answered why the mutations

occur mainly in these gene regions, how these mutations affect the onset of LHON

and if there are any other nuclear modifiers to these mtDNA mutations (Yen

et al. 2006).

Leber Hereditary Optic Neuropathy: A Mitochondrial Disease Unique in Many Ways 319

Page 12: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

5 Effect of Nuclear Genes on LHON

5.1 Chromosome X

The gender bias of LHON suggests the chromosome X genes may be involved in

this disease (Bu and Rotter 1991, 1992). According to the proposed two-locus

mitochondrial and X-chromosome-linked nuclear gene model (Bu and Rotter

1991, 1992), LHON symptoms occur only when the mtDNA pathogenic mutations

and chromosome X-sensitive gene(s) are present at the same time and the onset in

female carriers was dependent on the homozygous status of susceptible gene(s) in

chromosome X. This model is a reasonable explanation of the gender bias of

LHON, and different studies have identified several regions in chromosome X

showing linkage with LHON. The pioneer linkage study in six LHON families

performed by Vilkki et al. (1991) found that the onset of LHON is determined by

genes linked to the DXS7 region of chromosome X. The similar study performed by

Hudson et al. (2005) in 100 European LHON families identified that the DXS8090

(166)-DXS1068 (258) region of chromosome X is linked to the onset of LHON.

Also a study in a Brazilian LHON family found that region Xq25-Xp27.2 of

chromosome X confers susceptibility to LHON (Shankar et al. 2008). There were

relatively few studies performed in Chinese patients. Recently, Ji et al.

(2010) analyzed 12 microsatellite markers and 4 single nucleotide polymorphisms

(SNPs) in chromosome X in Chinese male LHON patients and controls and

identified two microsatellites (DXS6803 and DXS984) that are associated with

LHON. However, there were negative results reporting no association between

chromosome X and LHON (Man et al. 2002a; Oostra et al. 1996; Pegoraro

et al. 1996; Petruzzella et al. 2007). How genes in chromosome X affect LHON

is still a mystery. The recent study performed by Hudson et al. (2007a) indicated

that the oestrogen receptor-related genes in chromosome X may be susceptible

genes for LHON. These results need to be validated, and further functional charac-

terization should be carried out to elucidate the cause of the male-biased prevalence

of LHON.

5.2 Other Nuclear Factors

A nuclear genetic effect has been thought to account for the puzzle posed by the

incomplete penetrance of LHON, and identification of nuclear modifier(s) has

received wide attention from researchers in the field. A previous study focusing

on oxidative stress and apoptosis pathways identified EPHX1 and TP53 genes as

candidate nuclear genes associated with earlier onset of LHON (Ishikawa

et al. 2005). Recent genome-wide expression and genome-wide linkage studies

have further explored the potential nuclear genes that may play a modifying role in

LHON. In the genome-wide linkage study performed by Phasukkijwatana et al.

(2010) in patients from Thailand, chromosome region 3q26.2-3q28 was found to be

linked to LHON. This genomic region contains six genes, including PARL, OPA1

320 R. Bi et al.

Page 13: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

and others. Further analysis of SNPs in the PARL gene indicated that SNPs

rs3749446 and rs1402000 conferred a susceptibility to LHON (Phasukkijwatana

et al. 2010). However, the association between PARL SNPs and LHON could not be

replicated in the Han Chinese population in our validation study (Zhang

et al. 2010). The OPA1 gene is a pathogenic gene for autosomal dominant optic

atrophy (ADOA), which presents many overlapping clinical features with LHON

(Carelli et al. 2004). Previous studies have revealed that the OPA1 protein is located

in the mitochondrial inner membrane and plays important roles in mitochondrial

fusion and apoptosis (Olichon et al. 2003). Interestingly, PARL is also a mitochon-

drial protein and was recently found to regulate OPA1 in the apoptosis pathway

(Cipolat et al. 2006). In the genome-wide expression assay in LHON patients and

controls performed by Amero-Abu et al. (2010), 137 up-regulated and 152 down-

regulated genes were identified, in which the OPA1 gene was found to be signifi-

cantly down-regulated in LHON patients. Whether the OPA1-PARL pathway plays

a key role in the onset of optic atrophy, from either LHON or ADOA, is an

important and interesting question remaining to be answered.

A recent profiling of the mitochondrial proteome of fibroblasts from LHON

patients with m.11778G>A showed a down-regulation of bioenergetics and mito-

chondrial protein quality control pathways (Tun et al. 2014). Coincidentally,

Giordano et al. (2014) found that unaffected LHON mutation carriers had a

significantly higher mtDNA copy number and mitochondrial mass and higher

capacity for activating mitochondrial biogenesis under metabolic demand com-

pared with their affected maternally related relatives and healthy individuals. These

findings suggest that increased mitochondrial biogenesis in LHON mutation

carriers may overcome some of the pathogenic effect of the LHON mutation, and

this may account for the incomplete penetrance in LHON (Giordano et al. 2014).

These efforts aiming at identifying nuclear modifiers in LHON have provided some

helpful information for uncovering the puzzle of LHON, although we suggest that

more studies are essential to define the modifier genes and their regulatory

mechanisms in biochemical pathways.

6 Environmental Factors

The gender bias and incomplete penetrance of LHON strongly suggest that envi-

ronmental factors which are more specific to males than to females, such as

smoking and alcohol, may contribute to the increased disease risk of LHON in

males (Charlmers and Harding 1996; Kerrison et al. 2000; Kirkman et al. 2009). A

study by Charlmers and Harding (1996) showed that alcohol, but not smoking, may

significantly increase the penetrance of LHON. However, different observations

were obtained in a recent large-scale epidemiological study by Kirkman et al.

(2009), which showed that smoking is significantly linked to the penetrance of

LHON, independent of gender and alcohol intake, and excessive drinking would

lead to increased risk of LHON. Furthermore, a recent study found that oestrogens

could rescue the mitochondrial dysfunction caused by LHON primary mutations

Leber Hereditary Optic Neuropathy: A Mitochondrial Disease Unique in Many Ways 321

Page 14: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

(Giordano et al. 2011), which may underline the potential reason for the gender bias

of LHON. There are some anecdotal reports for the effect of exposure to toxic

substances or medicines, such as carbon monoxide poisoning (Hwang and Park

1996) and anti-tuberculosis medication including ethambutol (Seo et al. 2010), to

increase the clinical expression of LHON.

7 Biochemical Effects of mtDNA Mutations and TheirImplication for LHON Treatment

7.1 Biochemical Effect of mtDNA Mutations

It is of note that almost all the primary mutations and mutational hotspots for LHON

are located in genes that encode the subunits of complex I in the mitochondrial

respiration chain. For instance, mutations m.3460G>A (p.A52T) and m.3635G>A

(p.S110N) occur in the MT-ND1 subunit, mutation m.11778G>A (p.R340H) is

located in the MT-ND4 subunit and mutation m.14484T>C (p.M64V) is located in

the MT-ND6 subunit, whereas mutation m.10680G>A (p.A71T) is located in the

MT-ND4L subunit. All these mutations change the amino acids in different com-

plex I subunits and were considered to cause LHON through impairing the function

of complex I (Fig. 3) (Yen et al. 2006). Mitochondrial complex I, also known as

NADH dehydrogenase, is crucial for cellular metabolism and ATP production

(Hirst 2013). In addition, complex I is the major source of reactive oxygen species

(ROS) (Hirst 2013). A large number of studies have showed significantly decreased

complex I activity and respiration rate, as well as ATP level, in cybrids, fibroblasts

or lymphoblasts with LHON pathogenic mutations (Baracca et al. 2005; Brown

et al. 2000; Cock et al. 1999; Hirst 2013; Hofhaus et al. 1996; Lin et al. 2012;

Majander et al. 1996; Wong et al. 2002). It is suggested that the increased oxidative

stress caused by complex I dysfunction (Beretta et al. 2004; Wong et al. 2002) leads

to a series of mitochondrial defects and further triggers the clearance of damaged

mitochondria, which is also known as mitophagy (Melser et al. 2015). The excess of

mitophagy might be the major reason for the decreased level of mitochondrial mass,

as well as ATP production observed in LHON (Giordano et al. 2014). In this way

complex I dysfunction may eventually lead to increased apoptosis and cell death

(Hofhaus et al. 1996; Wong et al. 2002; Zanna et al. 2003) and be the potential

reason for the optic neuron loss observed in LHON patients and animal models

(Fig. 3). Consistent with this hypothesis, the chemically induced mouse model

initiated by the use of rotenone, an inhibitor of complex I, presented similar

degeneration of retinal ganglion cells as in LHON (Zhang et al. 2002). Therefore,

complex I dysfunction induced by LHON primary mutations may well be a key

pathogenic cause for the onset of LHON symptoms.

322 R. Bi et al.

Page 15: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

7.2 Functional Characterization of the LHON Primary Mutations

The pathogenicity of the LHON primary mutations needs to be proven by the use of

functional assays (Bandelt et al. 2009). One feasible approach is to develop cell

cybrids using blood platelets as these cells do not have nuclei (King and Attardi

1989), and this is now the most frequently used method to study the direct associa-

tion between mtDNA variants and alterations in mitochondrial function. Early

studies of the cybrids with LHON primary mutation m.11778G>A showed a

significantly decreased oxygen consumption, demonstrating a pathogenic role of

this mutation in impairing cell respiration (Vergani et al. 1995). A later study,

however, showed mutations m.3460G>A and m.11778G>A could result in com-

plex I defects, whereas m.14484T>C caused only a much milder biochemical

defect (Brown et al. 2000). In addition, LHON primary mutations were found to

disrupt glutamate transport in cybrid cell lines (Beretta et al. 2004).

Besides the trans-mitochondrial cybrids, the allotopic expression system, in

which the mtDNA gene was converted to a nuclear-encoded version and the protein

III III IV

V

ROS

G3460AG11778AT14484C

wild type

mtDNA

cytc

•apoptosis

•cell death

oxidative stress

Medicine

vitamin

idebenone

vitamin

idebenone

Fig. 3 Potential pathogenic role of LHON mutations in the cell and treatment strategy. The

dysfunction of complex I due to LHON mutations leads to the overproduction of ROS, and the

significantly increased oxidative stress triggers a series of mitochondrial-related dysfunctions and

increased apoptosis and cell death (Yen et al. 2006). Antioxidants like idebenone might therefore

be potentially effective medicines (Mashima et al. 2000). Gene therapy through allotopic expres-

sion of mitochondrial genes (Guy et al. 2002) or genome-editing technique using mitochondria-

targeted restriction endonucleases or TALENs (Reddy et al. 2015) may also increase the percent-

age of wild-type mtDNA and prevent the germline transmission of pathogenic mtDNA mutations

and is, therefore, another promising treatment for LHON symptoms

Leber Hereditary Optic Neuropathy: A Mitochondrial Disease Unique in Many Ways 323

Page 16: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

was first expressed in the cytosol and then imported into mitochondria with the

direction of mitochondrial targeting peptide (Fig. 3) (Manfredi et al. 2002), has also

been demonstrated to be efficient for the functional characterization of certain

mtDNA mutation(s) in our recent study (Bi et al. 2015) and others (Guy

et al. 2002; Manfredi et al. 2002). Through allotopic expression of wild-type ND4gene in cybrids with m.11778G>A, Guy et al. (2002) found that this method could

efficiently rescue the defective complex I-dependent ATP production and cell

growth in galactose medium. However, because of the high hydrophobicity of

mtDNA-encoded protein, the efficiency of this method was questioned in many

studies (Figueroa-Martinez et al. 2010; Oca-Cossio et al. 2003). Kaltimbacher et al.

(2006) optimized the allotopic expression system, in which the mRNA was first

targeted to the mitochondrial surface and then the protein translation was coupled

with protein transport. Allotopic expression of wild-type ND1 and ND4 genes in

fibroblasts containing m.3460G>A and m.11778G>A using this optimized method

successfully restored the complex I activity and mitochondrial dysfunction induced

by the LHON primary mutations (Bonnet et al. 2008). Using the same method,

Ellouze et al. (2008) introduced the ND4 gene harbouring the m.11778G>A

mutation into rat eyes by in vivo electroporation and developed an animal model

that mimics the essential aspects of LHON, including degeneration of retinal

ganglion cells and decline in visual performance. Further overexpression of the

wild-type ND4 gene in these LHON models could improve these signs.

7.3 New Avenues of Research: LHON Gene Therapy and GenomeEditing

In order to accomplish a more efficient gene delivery, the allotopic expression

method was further developed by injecting adeno-associated virus (AAV) with

mtDNA genes into eyes (Chadderton et al. 2013; Koilkonda et al. 2014; Yu

et al. 2012). Koilkonda et al. (2014) estimated the safety and effect of this AAV

mediate allotopic expression system in non-human primate and ex vivo human eyes

and found that the allotopic expression was efficient and long lasting with no

serious adverse reactions in most retinal ganglion cells. These studies indicated

that the gene therapy through allotopic expression of mitochondrial genes may be a

promising method in the treatment of LHON (Fig. 3). Also, Iyer et al. (2012) had

success by using healthy donor mtDNAs complexed with recombinant human

mitochondrial transcription factor A (TFAM) to improve respiration and biogenesis

in a LHON cell line. Recently, further work of this type has been reported from

France (Cwerman-Thibault et al. 2014; Cwerman-Thibault et al. 2015), and a trial

in carriers of the mutation m.11778G>A has been initiated. However, evidently,

there is still a long way to go before this method is fully accepted as a therapy.

Another promising approach for LHON therapy is mitochondrial replacement,

which involves spindle, pronuclear or polar body genome transfer into healthy

enucleated donor oocytes or embryos, and this approach has been successfully used

to eliminate pathogenic mtDNA variant(s) and to prevent the transmission of

324 R. Bi et al.

Page 17: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

mtDNA disease (Craven et al. 2010; Paull et al. 2013; Tachibana et al. 2009, 2013;

Wang et al. 2014). More recently, Reddy and colleagues (2015) took advantage of

the recently advanced genome-editing technique and selectively prevented the

germline transmission of a mtDNA haplotype using either mitochondria-targeted

restriction endonucleases or transcription activator-like effector nucleases

(TALENs). They successfully reduced LHON mutation level in mammalian

oocytes using mitochondria-targeted TALENs (mito-TALENs). We believe that

the most recently available CRISPR/Cas system, which is very powerful for

genome editing (Cong et al. 2013), will also be a workable method for mtDNA

genome editing in the future. The safety and efficacy of this mtDNA genome-

editing approach open a new avenue for LHON therapy (Fig. 3). However, there are

also potential concerns about the use of these techniques in a clinical setting

(Craven et al. 2011). Besides the complex technical issues, all these techniques

still need to be confirmed as being safe for use in humans.

7.4 Implications for Medical Therapy for LHON

There is currently no effective treatment that can be proven to prevent, or restore,

the visual loss in LHON (La Morgia et al. 2014; Yu-Wai-Man et al. 2014). But

several different approaches and therapies have been suggested. LHON mutation

carriers may have their symptoms precipitated by vitamin B12 deficiency, and

known carriers should have an adequate dietary intake of vitamin B12 (Pott and

Wong 2006). Spontaneous visual recovery is observed in some LHON patients, and

idebenone in combination with vitamin B2 and vitamin C has been suggested to

speed up this recovery (Fig. 3) (Mashima et al. 2000). Idebenone is described as an

antioxidant (Mordente et al. 1998) and transports electrons bypassing complex I to

complex III and thereby partly restores ATP production in cases where there is a

complex I defect (Giorgio et al. 2011; Haefeli et al. 2011). Pharmacological study

in a LHONmouse model with complex I dysfunction induced by rotenone indicated

that idebenone can restore the loss of retinal ganglion cells and vision defects in a

LHON mouse model (Heitz et al. 2012). In recent years, more evidence has shown

that LHON patients might benefit from idebenone treatment (Carelli et al. 2011;

Klopstock et al. 2011, 2013), suggesting that agents active against complex I

dysfunction and oxidative stress may be an efficient treatment for LHON in the

future (Fig. 3). However, the question of whether or not a new LHON patient should

be offered treatment with idebenone is still unproven.

Another approach to therapy for LHON is promising but still very much at the

theoretical stage and involves considering how to control the balance between

mitochondrial biogenesis and clearance (mitophagy). Mitochondrial biogenesis is

regulated by a complex system of transcriptional regulator networks of mitochon-

drial proteins (Vega et al. 2015). Furthermore, it was reported that the mitochon-

drial biogenesis and mitophagy could be regulated through the post-translational

modification of mitochondrial proteins (i.e. lysine acetylation), which was found to

be modified by the nutritional homeostasis (Webster et al. 2014). There are many

Leber Hereditary Optic Neuropathy: A Mitochondrial Disease Unique in Many Ways 325

Page 18: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

agents, chemicals and medicines that are known to reduce mitochondrial biogene-

sis, such as antibiotics targeting mitochondria like erythromycin, tetracycline,

chloramphenicol and glycylcyclines (Lamb et al. 2015). Recent studies have

identified several potentially therapeutic agents that could stimulate key regulators

of mitochondrial biogenesis (Sanchis-Gomar et al. 2014; Valero 2014), including

activators of the PPAR-PGC1α axis, bezafibrate, thiazolidinediones, pioglitazone,

rosiglitazone and fenofibrate (Uittenbogaard and Chiaramello 2014; Wu et al. 2014;

Yadav et al. 2014; Zamora and Villena 2014); agonists of AMPK, resveratrol,

AICAR and metformin (Uittenbogaard and Chiaramello 2014; Wu et al. 2014;

Zamora and Villena 2014); triggering factors of SIRT1, SRT1720, quercetin,

resveratrol and several isoflavone-derived compounds (Uittenbogaard and

Chiaramello 2014; Wu et al. 2014; Zamora and Villena 2014); and the agonists

of Nrf2, triterpenoids and Bacopa monnieri (Yadav et al. 2014). However, the

hypothesis to increase ATP production at times of stress through increasing mito-

chondrial mass has not been clinically proved. But it is an interesting line of

research that may be of significance to carriers of LHON mutations, as the possi-

bility of taking a simple medication to prevent the onset of symptoms is a pleasing

prospect.

8 Conclusion and Future Perspective

LHON, the most extensively studied mitochondrial disease, predominantly causes

blindness in young males and brings a lot of suffering to the patients and their

families. Although many primary mtDNA mutations have been identified, the

phenotypic expression of these primary mutations is very complex. Incomplete

disease penetrance, male-biased onset, involvement of additional genetic factors

(secondary mutations, mtDNA background, nuclear genetic modifiers, epigenetic

changes) or environmental factors (smoking, alcohol, toxic substance) are all

involved in the pathogenesis of the disorder; but their relevance has not been

fully understood. Currently, there is no effective treatment for LHON. Studies in

the past two decades have shown us that complex I dysfunction is the main

biochemical effect of the pathogenic mutations and gene therapy or medicine

aimed against complex I dysfunction are promising treatments for LHON. The

new techniques of genome editing also offer the possibility of a successful treat-

ment for LHON. Further studies focusing on the underlying mechanism of LHON

will be of importance in elucidating the pathophysiology of retinal neuron loss as

well as in searching for new clues that may finally prevent the visual loss caused

by LHON.

References

Abu-Amero KK, Jaber M, Hellani A, Bosley TM (2010) Genome-wide expression profile of

LHON patients with the 11778 mutation. Br J Ophthalmol 94:256–259

326 R. Bi et al.

Page 19: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

Achilli A, Perego UA, Bravi CM, Coble MD, Kong QP, Woodward SR, Salas A, Torroni A,

Bandelt HJ (2008) The phylogeny of the four pan-American MtDNA haplogroups:

implications for evolutionary and disease studies. PLoS One 3:e1764

Achilli A, Iommarini L, Olivieri A, Pala M, Hooshiar Kashani B, Reynier P, La Morgia C,

Valentino ML, Liguori R, Pizza F, Barboni P, Sadun F, De Negri AM, Zeviani M,

Dollfus H, Moulignier A, Ducos G, Orssaud C, Bonneau D, Procaccio V, Leo-Kottler B,

Fauser S, Wissinger B, Amati-Bonneau P, Torroni A, Carelli V (2012) Rare primary mito-

chondrial DNA mutations and probable synergistic variants in Leber’s hereditary optic neu-

ropathy. PLoS One 7:e42242

Bandelt HJ, Salas A, Taylor RW, Yao YG (2009) Exaggerated status of “novel” and “pathogenic”

mtDNA sequence variants due to inadequate database searches. Hum Mutat 30:191–196

Baracca A, Solaini G, Sgarbi G, Lenaz G, Baruzzi A, Schapira AH, Martinuzzi A, Carelli V (2005)

Severe impairment of complex I-driven adenosine triphosphate synthesis in Leber hereditary

optic neuropathy cybrids. Arch Neurol 62:730–736

Beretta S, Mattavelli L, Sala G, Tremolizzo L, Schapira AH, Martinuzzi A, Carelli V, Ferrarese C

(2004) Leber hereditary optic neuropathy mtDNA mutations disrupt glutamate transport in

cybrid cell lines. Brain 127:2183–2192

Bi R, Zhang AM, Yu D, Chen D, Yao YG (2010) Screening the three LHON primary mutations in

the general Chinese population by using an optimized multiplex allele-specific PCR. Clin

Chim Acta 411:1671–1674

Bi R, Zhang AM, Jia X, Zhang Q, Yao YG (2012) Complete mitochondrial DNA genome

sequence variation of Chinese families with mutation m.3635G>A and Leber hereditary

optic neuropathy. Mol Vis 18:3087–3094

Bi R, Zhang W, Yu D, Li X, Wang HZ, Hu QX, Zhang C, Lu W, Ni J, Fang Y, Li T, Yao YG

(2015) Mitochondrial DNA haplogroup B5 confers genetic susceptibility to Alzheimer’s

disease in Han Chinese. Neurobiol Aging 36:1604.e7

Black GC, Morten K, Laborde A, Poulton J (1996) Leber’s hereditary optic neuropathy:

heteroplasmy is likely to be significant in the expression of LHON in families with the 3460

ND1 mutation. Br J Ophthalmol 80:915–917

Bonnet C, Augustin S, Ellouze S, Benit P, Bouaita A, Rustin P, Sahel JA, Corral-Debrinski M

(2008) The optimized allotopic expression of ND1 or ND4 genes restores respiratory chain

complex I activity in fibroblasts harboring mutations in these genes. Biochim Biophys Acta

1783:1707–1717

Brown MD, Voljavec AS, Lott MT, Torroni A, Yang CC, Wallace DC (1992) Mitochondrial DNA

complex I and III mutations associated with Leber’s hereditary optic neuropathy. Genetics

130:163–173

BrownMD, Trounce IA, Jun AS, Allen JC, Wallace DC (2000) Functional analysis of lymphoblast

and cybrid mitochondria containing the 3460, 11778, or 14484 Leber’s hereditary optic

neuropathy mitochondrial DNA mutation. J Biol Chem 275:39831–39836

Brown MD, Allen JC, Van Stavern GP, Newman NJ, Wallace DC (2001a) Clinical, genetic, and

biochemical characterization of a Leber hereditary optic neuropathy family containing both the

11778 and 14484 primary mutations. Am J Med Genet 104:331–338

Brown MD, Zhadanov S, Allen JC, Hosseini S, Newman NJ, Atamonov VV, Mikhailovskaya IE,

Sukernik RI, Wallace DC (2001b) Novel mtDNA mutations and oxidative phosphorylation

dysfunction in Russian LHON families. Hum Genet 109:33–39

Bu XD, Rotter JI (1991) X chromosome-linked and mitochondrial gene control of Leber heredi-

tary optic neuropathy: evidence from segregation analysis for dependence on X chromosome

inactivation. Proc Natl Acad Sci U S A 88:8198–8202

Bu X, Rotter JI (1992) Leber hereditary optic neuropathy: estimation of number of embryonic

precursor cells and disease threshold in heterozygous affected females at the X-linked locus.

Clin Genet 42:143–148

Carelli V, Vergani L, Bernazzi B, Zampieron C, Bucchi L, Valentino M, Rengo C, Torroni A,

Martinuzzi A (2002) Respiratory function in cybrid cell lines carrying European mtDNA

Leber Hereditary Optic Neuropathy: A Mitochondrial Disease Unique in Many Ways 327

Page 20: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

haplogroups: implications for Leber’s hereditary optic neuropathy. Biochim Biophys Acta

1588:7–14

Carelli V, Ross-Cisneros FN, Sadun AA (2004) Mitochondrial dysfunction as a cause of optic

neuropathies. Prog Retin Eye Res 23:53–89

Carelli V, La Morgia C, Valentino ML, Rizzo G, Carbonelli M, De Negri AM, Sadun F, Carta A,

Guerriero S, Simonelli F, Sadun AA, Aggarwal D, Liguori R, Avoni P, Baruzzi A, Zeviani M,

Montagna P, Barboni P (2011) Idebenone treatment in Leber’s hereditary optic neuropathy.

Brain 134:e188

Chadderton N, Palfi A, Millington-Ward S, Gobbo O, Overlack N, Carrigan M, O’Reilly M,

Campbell M, Ehrhardt C, Wolfrum U, Humphries P, Kenna PF, Farrar GJ (2013) Intravitreal

delivery of AAV-NDI1 provides functional benefit in a murine model of Leber hereditary optic

neuropathy. Eur J Hum Genet 21:62–68

Charlmers RM, Harding AE (1996) A case-control study of Leber’s hereditary optic neuropathy.

Brain 119(Pt 5):1481–1486

Chinnery PF, Johnson MA, Wardell TM, Singh-Kler R, Hayes C, Brown DT, Taylor RW, Bindoff

LA, Turnbull DM (2000) The epidemiology of pathogenic mitochondrial DNAmutations. Ann

Neurol 48:188–193

Chinnery PF, Andrews RM, Turnbull DM, Howell NN (2001a) Leber hereditary optic neuropathy:

does heteroplasmy influence the inheritance and expression of the G11778A mitochondrial

DNA mutation? Am J Med Genet 98:235–243

Chinnery PF, Brown DT, Andrews RM, Singh-Kler R, Riordan-Eva P, Lindley J, Applegarth DA,

Turnbull DM, Howell N (2001b) The mitochondrial ND6 gene is a hot spot for mutations that

cause Leber’s hereditary optic neuropathy. Brain 124:209–218

Cipolat S, Rudka T, Hartmann D, Costa V, Serneels L, Craessaerts K, Metzger K, Frezza C,

Annaert W, D’Adamio L, Derks C, Dejaegere T, Pellegrini L, D’Hooge R, Scorrano L, De

Strooper B (2006) Mitochondrial rhomboid PARL regulates cytochrome c release during

apoptosis via OPA1-dependent cristae remodeling. Cell 126:163–175

Cock HR, Cooper JM, Schapira AH (1999) Functional consequences of the 3460-bp mitochondrial

DNA mutation associated with Leber’s hereditary optic neuropathy. J Neurol Sci 165:10–17

Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD, Wu X, Jiang W, Marraffini LA,

Zhang F (2013) Multiplex genome engineering using CRISPR/Cas systems. Science

339:819–823

Craven L, Tuppen HA, Greggains GD, Harbottle SJ, Murphy JL, Cree LM,Murdoch AP, Chinnery

PF, Taylor RW, Lightowlers RN, Herbert M, Turnbull DM (2010) Pronuclear transfer in

human embryos to prevent transmission of mitochondrial DNA disease. Nature 465:82–85

Craven L, Elson JL, Irving L, Tuppen HA, Lister LM, Greggains GD, Byerley S, Murdoch AP,

Herbert M, Turnbull D (2011) Mitochondrial DNA disease: new options for prevention. Hum

Mol Genet 20:R168–R174

Cwerman-Thibault H, Augustin S, Ellouze S, Sahel JA, Corral-Debrinski M (2014) Gene therapy

for mitochondrial diseases: Leber hereditary optic neuropathy as the first candidate for a

clinical trial. C R Biol 337:193–206

Cwerman-Thibault H, Augustin S, Lechauve C, Ayache J, Ellouze S, Sahel JA, Corral-Debrinski

M (2015) Nuclear expression of mitochondrial ND4 leads to the protein assembling in complex

I and prevents optic atrophy and visual loss. Mol Ther Methods Clin Dev 2:15003

DiMauro S, Schon EA (2003) Mitochondrial respiratory-chain diseases. N Engl J Med

348:2656–2668

Elliott HR, Samuels DC, Eden JA, Relton CL, Chinnery PF (2008) Pathogenic mitochondrial

DNA mutations are common in the general population. Am J Hum Genet 83:254–260

Ellouze S, Augustin S, Bouaita A, Bonnet C, Simonutti M, Forster V, Picaud S, Sahel JA, Corral-

Debrinski M (2008) Optimized allotopic expression of the human mitochondrial ND4 prevents

blindness in a rat model of mitochondrial dysfunction. Am J Hum Genet 83:373–387

Erickson RP (1972) Leber’s optic atrophy, a possible example of maternal inheritance. Am J Hum

Genet 24:348–349

328 R. Bi et al.

Page 21: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

Fauser S, Leo-Kottler B, Besch D, Luberichs J (2002a) Confirmation of the 14568 mutation in the

mitochondrial ND6 gene as causative in Leber’s hereditary optic neuropathy. Ophthalmic

Genet 23:191–197

Fauser S, Luberichs J, Besch D, Leo-Kottler B (2002b) Sequence analysis of the complete

mitochondrial genome in patients with Leber’s hereditary optic neuropathy lacking the three

most common pathogenic DNA mutations. Biochem Biophys Res Commun 295:342–347

Figueroa-Martinez F, Vazquez-Acevedo M, Cortes-Hernandez P, Garcia-Trejo JJ, Davidson E,

King MP, Gonzalez-Halphen D (2010) What limits the allotopic expression of nucleus-

encoded mitochondrial genes? The case of the chimeric Cox3 and Atp6 genes. Mitochondrion

11:147–154

Ghelli A, Porcelli AM, Zanna C, Vidoni S, Mattioli S, Barbieri A, Iommarini L, Pala M, Achilli A,

Torroni A, Rugolo M, Carelli V (2009) The background of mitochondrial DNA haplogroup J

increases the sensitivity of Leber’s hereditary optic neuropathy cells to 2,5-hexanedione

toxicity. PLoS One 4:e7922

Giordano C, Montopoli M, Perli E, Orlandi M, Fantin M, Ross-Cisneros FN, Caparrotta L,

Martinuzzi A, Ragazzi E, Ghelli A, Sadun AA, d’Amati G, Carelli V (2011) Oestrogens

ameliorate mitochondrial dysfunction in Leber’s hereditary optic neuropathy. Brain

134:220–234

Giordano C, Iommarini L, Giordano L, Maresca A, Pisano A, Valentino ML, Caporali L,

Liguori R, Deceglie S, Roberti M, Fanelli F, Fracasso F, Ross-Cisneros FN, D’Adamo P,

Hudson G, Pyle A, Yu-Wai-Man P, Chinnery PF, Zeviani M, Salomao SR, Berezovsky A,

Belfort R Jr, Ventura DF, Moraes M, Moraes Filho M, Barboni P, Sadun F, De Negri A, Sadun

AA, Tancredi A, Mancini M, d’Amati G, Loguercio Polosa P, Cantatore P, Carelli V (2014)

Efficient mitochondrial biogenesis drives incomplete penetrance in Leber’s hereditary optic

neuropathy. Brain 137:335–353

Giorgio V, Petronilli V, Ghelli A, Carelli V, Rugolo M, Lenaz G, Bernardi P (2011) The effects of

idebenone on mitochondrial bioenergetics. Biochim Biophys Acta 1817:363–369

Gropman A, Chen TJ, Perng CL, Krasnewich D, Chernoff E, Tifft C, Wong LJ (2004) Variable

clinical manifestation of homoplasmic G14459A mitochondrial DNA mutation. Am J Med

Genet A 124A:377–382

Guan MX (2011) Mitochondrial 12S rRNA mutations associated with aminoglycoside ototoxicity.

Mitochondrion 11:237–245

Guy J, Qi X, Pallotti F, Schon EA, Manfredi G, Carelli V, Martinuzzi A, Hauswirth WW, Lewin

AS (2002) Rescue of a mitochondrial deficiency causing Leber hereditary optic neuropathy.

Ann Neurol 52:534–542

Haefeli RH, Erb M, Gemperli AC, Robay D, Courdier Fruh I, Anklin C, Dallmann R, Gueven N

(2011) NQO1-dependent redox cycling of idebenone: effects on cellular redox potential and

energy levels. PLoS One 6:e17963

Harding AE, Sweeney MG, Govan GG, Riordan-Eva P (1995) Pedigree analysis in Leber heredi-

tary optic neuropathy families with a pathogenic mtDNAmutation. Am J HumGenet 57:77–86

Heitz FD, Erb M, Anklin C, Robay D, Pernet V, Gueven N (2012) Idebenone protects against

retinal damage and loss of vision in a mouse model of Leber’s hereditary optic neuropathy.

PLoS One 7:e45182

Hirst J (2013) Mitochondrial complex I. Annu Rev Biochem 82:551–575

Hofhaus G, Johns DR, Hurko O, Attardi G, Chomyn A (1996) Respiration and growth defects in

transmitochondrial cell lines carrying the 11778 mutation associated with Leber’s hereditary

optic neuropathy. J Biol Chem 271:13155–13161

Horvath J, Horvath R, Karcagi V, Komoly S, Johns DR (2002) Sequence analysis of Hungarian

LHON patients not carrying the common primary mutations. J Inherit Metab Dis 25:323–324

Howell N, Bindoff LA, McCullough DA, Kubacka I, Poulton J, Mackey D, Taylor L, Turnbull DM

(1991) Leber hereditary optic neuropathy: identification of the same mitochondrial ND1

mutation in six pedigrees. Am J Hum Genet 49:939–950

Leber Hereditary Optic Neuropathy: A Mitochondrial Disease Unique in Many Ways 329

Page 22: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

Hudson G, Keers S, Yu Wai Man P, Griffiths P, Huoponen K, Savontaus ML, Nikoskelainen E,

Zeviani M, Carrara F, Horvath R, Karcagi V, Spruijt L, de Coo IF, Smeets HJ, Chinnery PF

(2005) Identification of an X-chromosomal locus and haplotype modulating the phenotype of a

mitochondrial DNA disorder. Am J Hum Genet 77:1086–1091

Hudson G, Carelli V, Horvath R, Zeviani M, Smeets HJ, Chinnery PF (2007a) X-Inactivation

patterns in females harboring mtDNA mutations that cause Leber hereditary optic neuropathy.

Mol Vis 13:2339–2343

Hudson G, Carelli V, Spruijt L, Gerards M, Mowbray C, Achilli A, Pyle A, Elson J, Howell N, La

Morgia C, Valentino ML, Huoponen K, Savontaus ML, Nikoskelainen E, Sadun AA, Salomao

SR, Belfort R Jr, Griffiths P, Man PY, de Coo RF, Horvath R, Zeviani M, Smeets HJ,

Torroni A, Chinnery PF (2007b) Clinical expression of Leber hereditary optic neuropathy is

affected by the mitochondrial DNA-haplogroup background. Am J Hum Genet 81:228–233

Huoponen K, Vilkki J, Aula P, Nikoskelainen EK, Savontaus ML (1991) A new mtDNA mutation

associated with Leber hereditary optic neuroretinopathy. Am J Hum Genet 48:1147–1153

Huoponen K, Lamminen T, Juvonen V, Aula P, Nikoskelainen E, Savontaus ML (1993) The

spectrum of mitochondrial DNA mutations in families with Leber hereditary optic

neuroretinopathy. Hum Genet 92:379–384

Hwang JM, Park HW (1996) Carbon monoxide poisoning as an epigenetic factor for Leber’s

hereditary optic neuropathy. Korean J Ophthalmol 10:122–123

Ishikawa K, Funayama T, Ohde H, Inagaki Y, Mashima Y (2005) Genetic variants of TP53 and

EPHX1 in Leber’s hereditary optic neuropathy and their relationship to age at onset. Jpn J

Ophthalmol 49:121–126

Iyer S, Bergquist K, Young K, Gnaiger E, Rao RR, Bennett JP Jr (2012) Mitochondrial gene

therapy improves respiration, biogenesis, and transcription in G11778A Leber’s hereditary

optic neuropathy and T8993G Leigh’s syndrome cells. Hum Gene Ther 23:647–657

Jacobi FK, Leo-Kottler B, Mittelviefhaus K, Zrenner E, Meyer J, Pusch CM, Wissinger B (2001)

Segregation patterns and heteroplasmy prevalence in Leber’s hereditary optic neuropathy.

Invest Ophthalmol Vis Sci 42:1208–1214

Jaros E, Mahad DJ, Hudson G, Birchall D, Sawcer SJ, Griffiths PG, Sunter J, Compston DA, Perry

RH, Chinnery PF (2007) Primary spinal cord neurodegeneration in Leber hereditary optic

neuropathy. Neurology 69:214–216

Ji Y, Zhang AM, Jia X, Zhang YP, Xiao X, Li S, Guo X, Bandelt HJ, Zhang Q, Yao YG (2008)

Mitochondrial DNA haplogroups M7b102 and M8a affect clinical expression of Leber heredi-

tary optic neuropathy in Chinese families with the m.11778G>A mutation. Am J Hum Genet

83:760–768

Ji Y, Jia X, Li S, Xiao X, Guo X, Zhang Q (2010) Evaluation of the X-linked modifier loci for

Leber hereditary optic neuropathy with the G11778A mutation in Chinese. Mol Vis

16:416–424

Jia X, Li S, Xiao X, Guo X, Zhang Q (2006) Molecular epidemiology of mtDNA mutations in

903 Chinese families suspected with Leber hereditary optic neuropathy. J Hum Genet

51:851–856

Jia X, Li S, Wang P, Guo X, Zhang Q (2010) mtDNAm.3635G>Amay be classified as a common

primary mutation for Leber hereditary optic neuropathy in the Chinese population. Biochem

Biophys Res Commun 403:237–241

Kaewsutthi S, Phasukkijwatana N, Joyjinda Y, Chuenkongkaew W, Kunhapan B, Tun AW,

Suktitipat B, Lertrit P (2011) Mitochondrial haplogroup background may influence Southeast

Asian G11778A Leber hereditary optic neuropathy. Invest Ophthalmol Vis Sci 52:4742–4748

Kaltimbacher V, Bonnet C, Lecoeuvre G, Forster V, Sahel JA, Corral-Debrinski M (2006) mRNA

localization to the mitochondrial surface allows the efficient translocation inside the organelle

of a nuclear recoded ATP6 protein. RNA 12:1408–1417

Kerrison JB, Miller NR, Hsu F, Beaty TH, Maumenee IH, Smith KH, Savino PJ, Stone EM,

Newman NJ (2000) A case-control study of tobacco and alcohol consumption in Leber

hereditary optic neuropathy. Am J Ophthalmol 130:803–812

330 R. Bi et al.

Page 23: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

Khan NA, Govindaraj P, Jyothi V, Meena AK, Thangaraj K (2013) Co-occurrence of m.1555A>G

and m.11778G>A mitochondrial DNA mutations in two Indian families with strikingly

different clinical penetrance of Leber hereditary optic neuropathy. Mol Vis 19:1282–1289

Kim JY, Hwang JM, Park SS (2002) Mitochondrial DNA C4171A/ND1 is a novel primary

causative mutation of Leber’s hereditary optic neuropathy with a good prognosis. Ann Neurol

51:630–634

King MP, Attardi G (1989) Human cells lacking mtDNA: repopulation with exogenous

mitochondria by complementation. Science 246:500–503

Kirkman MA, Yu-Wai-Man P, Korsten A, Leonhardt M, Dimitriadis K, De Coo IF, Klopstock T,

Chinnery PF (2009) Gene-environment interactions in Leber hereditary optic neuropathy.

Brain 132:2317–2326

Klopstock T, Yu-Wai-Man P, Dimitriadis K, Rouleau J, Heck S, Bailie M, Atawan A,

Chattopadhyay S, Schubert M, Garip A, Kernt M, Petraki D, Rummey C, Leinonen M,

Metz G, Griffiths PG, Meier T, Chinnery PF (2011) A randomized placebo-controlled trial

of idebenone in Leber’s hereditary optic neuropathy. Brain 134:2677–2686

Klopstock T, Metz G, Yu-Wai-Man P, Buchner B, Gallenmuller C, Bailie M, Nwali N, Griffiths

PG, von Livonius B, Reznicek L, Rouleau J, Coppard N, Meier T, Chinnery PF (2013)

Persistence of the treatment effect of idebenone in Leber’s hereditary optic neuropathy.

Brain 136:e230

Kodron A, Krawczynski MR, Tonska K, Bartnik E (2014) m.3635G>A mutation as a cause of

Leber hereditary optic neuropathy. J Clin Pathol 67:639–641

Koilkonda RD, Yu H, Chou TH, Feuer WJ, Ruggeri M, Porciatti V, Tse D, Hauswirth WW,

Chiodo V, Boye SL, Lewin AS, Neuringer M, Renner L, Guy J (2014) Safety and effects of the

vector for the Leber hereditary optic neuropathy gene therapy clinical trial. JAMA Ophthalmol

132:409–420

Kokotas H, Petersen MB, Willems PJ (2007) Mitochondrial deafness. Clin Genet 71:379–391

Kong QP, Bandelt HJ, Sun C, Yao YG, Salas A, Achilli A, Wang CY, Zhong L, Zhu CL, Wu SF,

Torroni A, Zhang YP (2006) Updating the East Asian mtDNA phylogeny: a prerequisite for the

identification of pathogenic mutations. Hum Mol Genet 15:2076–2086

Kong Q-P, Bandelt H-J, Zhao M, Zhang Y-P, Yao Y-G (2010) Reply to van Oven: suggestions and

caveats for naming mtDNA haplogroup. Proc Natl Acad Sci U S A 107:E40–E41

La Morgia C, Achilli A, Iommarini L, Barboni P, Pala M, Olivieri A, Zanna C, Vidoni S, Tonon C,

Lodi R, Vetrugno R, Mostacci B, Liguori R, Carroccia R, Montagna P, Rugolo M, Torroni A,

Carelli V (2008) Rare mtDNA variants in Leber hereditary optic neuropathy families with

recurrence of myoclonus. Neurology 70:762–770

La Morgia C, Carbonelli M, Barboni P, Sadun AA, Carelli V (2014) Medical management of

hereditary optic neuropathies. Front Neurol 5:141

Lamb R, Ozsvari B, Lisanti CL, Tanowitz HB, Howell A, Martinez-Outschoorn UE, Sotgia F,

Lisanti MP (2015) Antibiotics that target mitochondria effectively eradicate cancer stem cells,

across multiple tumor types: treating cancer like an infectious disease. Oncotarget

6:4569–4584

Leber T (1871) Ueber hereditareund congenital angelegte sehnervenleiden. Graefes Arch Clin Exp

Ophthalmol 17:249–291

Lin CS, Sharpley MS, Fan W, Waymire KG, Sadun AA, Carelli V, Ross-Cisneros FN, Baciu P,

Sung E, McManus MJ, Pan BX, Gil DW, Macgregor GR, Wallace DC (2012) Mouse mtDNA

mutant model of Leber hereditary optic neuropathy. Proc Natl Acad Sci U S A

109:20065–20070

Mackey D, Howell N (1992) A variant of Leber hereditary optic neuropathy characterized by

recovery of vision and by an unusual mitochondrial genetic etiology. Am J Hum Genet

51:1218–1228

Mackey DA, Oostra RJ, Rosenberg T, Nikoskelainen E, Bronte-Stewart J, Poulton J, Harding AE,

Govan G, Bolhuis PA, Norby S (1996) Primary pathogenic mtDNA mutations in

Leber Hereditary Optic Neuropathy: A Mitochondrial Disease Unique in Many Ways 331

Page 24: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

multigeneration pedigrees with Leber hereditary optic neuropathy. Am J Hum Genet

59:481–485

Macmillan C, Kirkham T, Fu K, Allison V, Andermann E, Chitayat D, Fortier D, Gans M, Hare H,

Quercia N, Zackon D, Shoubridge EA (1998) Pedigree analysis of French Canadian families

with T14484C Leber’s hereditary optic neuropathy. Neurology 50:417–422

Macmillan C, Johns TA, Fu K, Shoubridge EA (2000) Predominance of the T14484C mutation in

French-Canadian families with Leber hereditary optic neuropathy is due to a founder effect.

Am J Hum Genet 66:332–335

Majander A, Finel M, Savontaus ML, Nikoskelainen E, Wikstrom M (1996) Catalytic activity of

complex I in cell lines that possess replacement mutations in the ND genes in Leber’s

hereditary optic neuropathy. Eur J Biochem 239:201–207

Man PY, Brown DT, Wehnert MS, Zeviani M, Carrara F, Turnbull DM, Chinnery PF (2002a)

NDUFA-1 is not a nuclear modifier gene in Leber hereditary optic neuropathy. Neurology

58:1861–1862

Man PY, Turnbull DM, Chinnery PF (2002b) Leber hereditary optic neuropathy. J Med Genet

39:162–169

Man PY, Griffiths PG, Brown DT, Howell N, Turnbull DM, Chinnery PF (2003) The epidemiol-

ogy of Leber hereditary optic neuropathy in the North East of England. Am J Hum Genet

72:333–339

Manfredi G, Fu J, Ojaimi J, Sadlock JE, Kwong JQ, Guy J, Schon EA (2002) Rescue of a

deficiency in ATP synthesis by transfer of MTATP6, a mitochondrial DNA-encoded gene, to

the nucleus. Nat Genet 30:394–399

Mashima Y, Kigasawa K, Wakakura M, Oguchi Y (2000) Do idebenone and vitamin therapy

shorten the time to achieve visual recovery in Leber hereditary optic neuropathy? J

Neuroophthalmol 20:166–170

Melser S, Lavie J, Benard G (2015) Mitochondrial degradation and energy metabolism. Biochim

Biophys Acta 1853:2812–2821

Mimaki M, Ikota A, Sato A, Komaki H, Akanuma J, Nonaka I, Goto Y (2003) A double mutation

(G11778A and G12192A) in mitochondrial DNA associated with Leber’s hereditary optic

neuropathy and cardiomyopathy. J Hum Genet 48:47–50

Mordente A, Martorana GE, Minotti G, Giardina B (1998) Antioxidant properties of

2,3-dimethoxy-5-methyl-6-(10-hydroxydecyl)-1,4-benzoquinone (idebenone). Chem Res

Toxicol 11:54–63

Nikoskelainen EK, Huoponen K, Juvonen V, Lamminen T, Nummelin K, Savontaus ML (1996)

Ophthalmologic findings in Leber hereditary optic neuropathy, with special reference to

mtDNA mutations. Ophthalmology 103:504–514

Oca-Cossio J, Kenyon L, Hao H, Moraes CT (2003) Limitations of allotopic expression of

mitochondrial genes in mammalian cells. Genetics 165:707–720

Olichon A, Baricault L, Gas N, Guillou E, Valette A, Belenguer P, Lenaers G (2003) Loss of

OPA1 perturbates the mitochondrial inner membrane structure and integrity, leading to

cytochrome c release and apoptosis. J Biol Chem 278:7743–7746

Oostra RJ, Kemp S, Bolhuis PA, Bleeker-Wagemakers EM (1996) No evidence for ‘skewed’

inactivation of the X-chromosome as cause of Leber’s hereditary optic neuropathy in female

carriers. Hum Genet 97:500–505

Pakendorf B, Stoneking M (2005) Mitochondrial DNA and human evolution. Annu Rev Genomics

Hum Genet 6:165–183

Paull D, Emmanuele V, Weiss KA, Treff N, Stewart L, Hua H, Zimmer M, Kahler DJ, Goland RS,

Noggle SA, Prosser R, Hirano M, Sauer MV, Egli D (2013) Nuclear genome transfer in human

oocytes eliminates mitochondrial DNA variants. Nature 493:632–637

Pegoraro E, Carelli V, Zeviani M, Cortelli P, Montagna P, Barboni P, Angelini C, Hoffman EP

(1996) X-inactivation patterns in female Leber’s hereditary optic neuropathy patients do not

support a strong X-linked determinant. Am J Med Genet 61:356–362

332 R. Bi et al.

Page 25: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

Petit C, Levilliers J, Hardelin JP (2001) Molecular genetics of hearing loss. Annu Rev Genet

35:589–646

Petruzzella V, Tessa A, Torraco A, Fattori F, Dotti MT, Bruno C, Cardaioli E, Papa S, Federico A,

Santorelli FM (2007) The NDUFB11 gene is not a modifier in Leber hereditary optic neuropa-

thy. Biochem Biophys Res Commun 355:181–187

Phasukkijwatana N, Chuenkongkaew WL, Suphavilai R, Suktitipat B, Pingsuthiwong S,

Ruangvaravate N, Atchaneeyasakul LO, Warrasak S, Poonyathalang A, Sura T, Lertrit P

(2006) The unique characteristics of Thai Leber hereditary optic neuropathy: analysis of

30 G11778A pedigrees. J Hum Genet 51:298–304

Phasukkijwatana N, Kunhapan B, Stankovich J, Chuenkongkaew WL, Thomson R, Thornton T,

Bahlo M, Mushiroda T, Nakamura Y, Mahasirimongkol S, Tun AW, Srisawat C,

Limwongse C, Peerapittayamongkol C, Sura T, Suthammarak W, Lertrit P (2010) Genome-

wide linkage scan and association study of PARL to the expression of LHON families in

Thailand. Hum Genet 128:39–49

Pott JW, Wong KH (2006) Leber’s hereditary optic neuropathy and vitamin B12 deficiency.

Graefes Arch Clin Exp Ophthalmol 244:1357–1359

Puomila A, Hamalainen P, Kivioja S, Savontaus ML, Koivumaki S, Huoponen K, Nikoskelainen E

(2007) Epidemiology and penetrance of Leber hereditary optic neuropathy in Finland. Eur J

Hum Genet 15:1079–1089

Reddy P, Ocampo A, Suzuki K, Luo J, Bacman SR, Williams SL, Sugawara A, Okamura D,

Tsunekawa Y, Wu J, Lam D, Xiong X, Montserrat N, Esteban CR, Liu GH, Sancho-Martinez I,

Manau D, Civico S, Cardellach F, Del Mar O’Callaghan M, Campistol J, Zhao H, Campistol

JM, Moraes CT, Izpisua Belmonte JC (2015) Selective elimination of mitochondrial mutations

in the germline by genome editing. Cell 161:459–469

Riordan-Eva P, Harding AE (1995) Leber’s hereditary optic neuropathy: the clinical relevance of

different mitochondrial DNA mutations. J Med Genet 32:81–87

Sadun AA, Carelli V, Salomao SR, Berezovsky A, Quiros P, Sadun F, DeNegri AM, Andrade R,

Schein S, Belfort R (2002) A very large Brazilian pedigree with 11778 Leber’s hereditary optic

neuropathy. Trans Am Ophthalmol Soc 100:169–178

Sanchis-Gomar F, Garcia-Gimenez JL, Gomez-Cabrera MC, Pallardo FV (2014) Mitochondrial

biogenesis in health and disease. Molecular and therapeutic approaches. Curr Pharm Des

20:5619–5633

Seedorff T (1985) The inheritance of Leber’s disease. A genealogical follow-up study. Acta

Ophthalmol (Copenh) 63:135–145

Seo JH, Hwang JM, Park SS (2010) Antituberculosis medication as a possible epigenetic factor of

Leber’s hereditary optic neuropathy. Clin Experiment Ophthalmol 38:363–366

Shafa Shariat Panahi M, Houshmand M, Tabassi AR (2006) Mitochondrial D-loop variation in

Leber hereditary neuropathy patients harboring primary G11778A, G3460A, T14484C

mutations: J and W haplogroups as high-risk factors. Arch Med Res 37:1028–1033

Shankar SP, Fingert JH, Carelli V, Valentino ML, King TM, Daiger SP, Salomao SR,

Berezovsky A, Belfort R Jr, Braun TA, Sheffield VC, Sadun AA, Stone EM (2008) Evidence

for a novel x-linked modifier locus for Leber hereditary optic neuropathy. Ophthalmic Genet

29:17–24

Shoffner JM, Brown MD, Stugard C, Jun AS, Pollock S, Haas RH, Kaufman A, Koontz D, Kim Y,

Graham JR et al (1995) Leber’s hereditary optic neuropathy plus dystonia is caused by a

mitochondrial DNA point mutation. Ann Neurol 38:163–169

Sorajja P, Sweeney MG, Chalmers R, Sachdev B, Syrris P, Hanna M, Wood ND, McKenna WJ,

Elliott PM (2003) Cardiac abnormalities in patients with Leber’s hereditary optic neuropathy.

Heart 89:791–792

Spruijt L, Kolbach DN, de Coo RF, Plomp AS, Bauer NJ, Smeets HJ, de Die-Smulders CE (2006)

Influence of mutation type on clinical expression of Leber hereditary optic neuropathy. Am J

Ophthalmol 141:676–682

Leber Hereditary Optic Neuropathy: A Mitochondrial Disease Unique in Many Ways 333

Page 26: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

Suissa S, Wang Z, Poole J, Wittkopp S, Feder J, Shutt TE, Wallace DC, Shadel GS, Mishmar D

(2009) Ancient mtDNA genetic variants modulate mtDNA transcription and replication. PLoS

Genet 5:e1000474

Tachibana M, Sparman M, Sritanaudomchai H, Ma H, Clepper L, Woodward J, Li Y, Ramsey C,

Kolotushkina O, Mitalipov S (2009) Mitochondrial gene replacement in primate offspring and

embryonic stem cells. Nature 461:367–372

Tachibana M, Amato P, Sparman M, Woodward J, Sanchis DM, Ma H, Gutierrez NM, Tippner-

Hedges R, Kang E, Lee HS, Ramsey C, Masterson K, Battaglia D, Lee D, Wu D, Jensen J,

Patton P, Gokhale S, Stouffer R, Mitalipov S (2013) Towards germline gene therapy of

inherited mitochondrial diseases. Nature 493:627–631

Tonska K, Kurzawa M, Ambroziak AM, Korwin-Rujna M, Szaflik JP, Grabowska E, Szaflik J,

Bartnik E (2008) A family with 3460G>A and 11778G>A mutations and haplogroup analysis

of Polish Leber hereditary optic neuropathy patients. Mitochondrion 8:383–388

Torroni A, Petrozzi M, D’Urbano L, Sellitto D, Zeviani M, Carrara F, Carducci C, Leuzzi V,

Carelli V, Barboni P, De Negri A, Scozzari R (1997) Haplotype and phylogenetic analyses

suggest that one European-specific mtDNA background plays a role in the expression of Leber

hereditary optic neuropathy by increasing the penetrance of the primary mutations 11778 and

14484. Am J Hum Genet 60:1107–1121

Torroni A, Richards M, Macaulay V, Forster P, Villems R, Norby S, Savontaus ML, Huoponen K,

Scozzari R, Bandelt HJ (2000) mtDNA haplogroups and frequency patterns in Europe. Am J

Hum Genet 66:1173–1177

Tun AW, Chaiyarit S, Kaewsutthi S, Katanyoo W, Chuenkongkaew W, Kuwano M, Tomonaga T,

Peerapittayamongkol C, Thongboonkerd V, Lertrit P (2014) Profiling the mitochondrial

proteome of Leber’s Hereditary Optic Neuropathy (LHON) in Thailand: down-regulation of

bioenergetics and mitochondrial protein quality control pathways in fibroblasts with the

11778G>A mutation. PLoS One 9:e106779

Uittenbogaard M, Chiaramello A (2014) Mitochondrial biogenesis: a therapeutic target for

neurodevelopmental disorders and neurodegenerative diseases. Curr Pharm Des 20:5574–5593

Valentino ML, Barboni P, Ghelli A, Bucchi L, Rengo C, Achilli A, Torroni A, Lugaresi A, Lodi R,

Barbiroli B, Dotti M, Federico A, Baruzzi A, Carelli V (2004) The ND1 gene of complex I is a

mutational hot spot for Leber’s hereditary optic neuropathy. Ann Neurol 56:631–641

Valero T (2014) Mitochondrial biogenesis: pharmacological approaches. Curr Pharm Des

20:5507–5509

van Oven M, Kayser M (2009) Updated comprehensive phylogenetic tree of global human

mitochondrial DNA variation. Hum Mutat 30:E386–E394

Vega RB, Horton JL, Kelly DP (2015) Maintaining ancient organelles: mitochondrial biogenesis

and maturation. Circ Res 116:1820–1834

Vergani L, Martinuzzi A, Carelli V, Cortelli P, Montagna P, Schievano G, Carrozzo R, Angelini C,

Lugaresi E (1995) MtDNA mutations associated with Leber’s hereditary optic neuropathy:

studies on cytoplasmic hybrid (cybrid) cells. Biochem Biophys Res Commun 210:880–888

Vilkki J, Ott J, Savontaus ML, Aula P, Nikoskelainen EK (1991) Optic atrophy in Leber hereditary

optic neuroretinopathy is probably determined by an X-chromosomal gene closely linked to

DXS7. Am J Hum Genet 48:486–491

Wallace DC, Singh G, Lott MT, Hodge JA, Schurr TG, Lezza AM, Elsas LJ 2nd, Nikoskelainen

EK (1988) Mitochondrial DNA mutation associated with Leber’s hereditary optic neuropathy.

Science 242:1427–1430

Wang HW, Jia X, Ji Y, Kong QP, Zhang Q, Yao YG, Zhang YP (2008) Strikingly different

penetrance of LHON in two Chinese families with primary mutation G11778A is independent

of mtDNA haplogroup background and secondary mutation G13708A. Mutat Res 643:48–53

Wang T, Sha H, Ji D, Zhang HL, Chen D, Cao Y, Zhu J (2014) Polar body genome transfer for

preventing the transmission of inherited mitochondrial diseases. Cell 157:1591–1604

Webster BR, Scott I, Traba J, Han K, Sack MN (2014) Regulation of autophagy and mitophagy by

nutrient availability and acetylation. Biochim Biophys Acta 1841:525–534

334 R. Bi et al.

Page 27: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

Wissinger B, Besch D, Baumann B, Fauser S, Christ-Adler M, Jurklies B, Zrenner E, Leo-Kottler

B (1997) Mutation analysis of the ND6 gene in patients with Lebers hereditary optic neuropa-

thy. Biochem Biophys Res Commun 234:511–515

Wong A, Cavelier L, Collins-Schramm HE, Seldin MF, McGrogan M, Savontaus ML, Cortopassi

GA (2002) Differentiation-specific effects of LHON mutations introduced into neuronal NT2

cells. Hum Mol Genet 11:431–438

Wu YT, Wu SB, Wei YH (2014) Metabolic reprogramming of human cells in response to

oxidative stress: implications in the pathophysiology and therapy of mitochondrial diseases.

Curr Pharm Des 20:5510–5526

Yadav A, Agarwal S, Tiwari SK, Chaturvedi RK (2014) Mitochondria: prospective targets for

neuroprotection in Parkinson’s disease. Curr Pharm Des 20:5558–5573

Yang J, Zhu Y, Tong Y, Chen L, Liu L, Zhang Z, Wang X, Huang D, Qiu W, Zhuang S, Ma X

(2009a) Confirmation of the mitochondrial ND1 gene mutation G3635A as a primary LHON

mutation. Biochem Biophys Res Commun 386:50–54

Yang J, Zhu Y, Tong Y, Zhang Z, Chen L, Chen S, Cao Z, Liu C, Xu J, Ma X (2009b) The novel

G10680A mutation is associated with complete penetrance of the LHON/T14484C family.

Mitochondrion 9:273–278

Yao YG, Kajigaya S, Young NS (2015) Mitochondrial DNA mutations in single human blood

cells. Mutat Res 779:68–77

Yen MY,Wang AG, Wei YH (2006) Leber’s hereditary optic neuropathy: a multifactorial disease.

Prog Retin Eye Res 25:381–396

Yu D, Jia X, Zhang AM, Guo X, Zhang YP, Zhang Q, Yao YG (2010a) Molecular characterization

of six Chinese families with m.3460G>A and Leber hereditary optic neuropathy.

Neurogenetics 11:349–356

Yu D, Jia X, Zhang AM, Li S, Zou Y, Zhang Q, Yao YG (2010b) Mitochondrial DNA sequence

variation and haplogroup distribution in Chinese patients with LHON and m.14484T>C. PLoS

One 5:e13426

Yu H, Koilkonda RD, Chou TH, Porciatti V, Ozdemir SS, Chiodo V, Boye SL, Boye SE,

Hauswirth WW, Lewin AS, Guy J (2012) Gene delivery to mitochondria by targeting modified

adenoassociated virus suppresses Leber’s hereditary optic neuropathy in a mouse model. Proc

Natl Acad Sci U S A 109:E1238–E1247

Yu-Wai-Man P, Griffiths PG, Hudson G, Chinnery PF (2009) Inherited mitochondrial optic

neuropathies. J Med Genet 46:145–158

Yu-Wai-Man P, Votruba M, Moore AT, Chinnery PF (2014) Treatment strategies for inherited

optic neuropathies: past, present and future. Eye (Lond) 28:521–537

Zamora M, Villena JA (2014) Targeting mitochondrial biogenesis to treat insulin resistance. Curr

Pharm Des 20:5527–5557

Zanna C, Ghelli A, Porcelli AM, Carelli V, Martinuzzi A, Rugolo M (2003) Apoptotic cell death of

cybrid cells bearing Leber’s hereditary optic neuropathy mutations is caspase independent.

Ann N Y Acad Sci 1010:213–217

Zhang X, Jones D, Gonzalez-Lima F (2002) Mouse model of optic neuropathy caused by

mitochondrial complex I dysfunction. Neurosci Lett 326:97–100

Zhang AM, Jia X, Yao YG, Zhang Q (2008) Co-occurrence of A1555G and G11778A in a Chinese

family with high penetrance of Leber’s hereditary optic neuropathy. Biochem Biophys Res

Commun 376:221–224

Zhang AM, Zou Y, Guo X, Jia X, Zhang Q, Yao YG (2009) Mitochondrial DNA mutation

m.3635G>A may be associated with Leber hereditary optic neuropathy in Chinese. Biochem

Biophys Res Commun 386:392–395

Zhang AM, Jia X, Zhang Q, Yao YG (2010) No association between the SNPs (rs3749446 and

rs1402000) in the PARL gene and LHON in Chinese patients with m.11778G>A. Hum Genet

128:465–468

Leber Hereditary Optic Neuropathy: A Mitochondrial Disease Unique in Many Ways 335

Page 28: Leber Hereditary Optic Neuropathy: A Mitochondrial Disease …. Exp. Pharmacol.pdf · 2017-10-10 · Division of Medical Genetics & Evolutionary Medicine, Key Laboratory of Animal

Zhang AM, Bandelt HJ, Jia X, Zhang W, Li S, Yu D, Wang D, Zhuang XY, Zhang Q, Yao YG

(2011a) Is mitochondrial tRNA(phe) variant m.593T>C a synergistically pathogenic mutation

in Chinese LHON families with m.11778G>A? PLoS One 6:e26511

Zhang AM, Jia X, Bi R, Salas A, Li S, Xiao X, Wang P, Guo X, Kong QP, Zhang Q, Yao YG

(2011b) Mitochondrial DNA haplogroup background affects LHON, but not suspected LHON,

in Chinese patients. PLoS One 6:e27750

Zhang AM, Jia X, Guo X, Zhang Q, Yao YG (2012) Mitochondrial DNA mutation m.10680G>A

is associated with Leber hereditary optic neuropathy in Chinese patients. J Transl Med 10:43

Zhang J, Jiang P, Jin X, Liu X, Zhang M, Xie S, Gao M, Zhang S, Sun YH, Zhu J, Ji Y, Wei QP,

Tong Y, Guan MX (2014) Leber’s hereditary optic neuropathy caused by the homoplasmic

ND1 m.3635G>A mutation in nine Han Chinese families. Mitochondrion 18:18–26

Zou Y, Jia X, Zhang AM, Wang WZ, Li S, Guo X, Kong QP, Zhang Q, Yao YG (2010) The

MT-ND1 and MT-ND5 genes are mutational hotspots for Chinese families with clinical

features of LHON but lacking the three primary mutations. Biochem Biophys Res Commun

399:179–185

336 R. Bi et al.